MAGnesium-oral supplementation to reduce PAin in patients with severe PERipheral arterial occlusive disease: the MAG-PAPER randomised clinical trial protocol

Monica Aida Venturini, Sergio Zappa, Cosetta Minelli, Stefano Bonardelli, Laura Lamberti, Luca Bisighini, Marta Zangrandi, Maddalena Turin, Francesco Rizzo, Andrea Rizzolo, Nicola Latronico, Monica Aida Venturini, Sergio Zappa, Cosetta Minelli, Stefano Bonardelli, Laura Lamberti, Luca Bisighini, Marta Zangrandi, Maddalena Turin, Francesco Rizzo, Andrea Rizzolo, Nicola Latronico

Abstract

Introduction: Magnesium exerts analgaesic effects in several animal pain models, as well as in patients affected by acute postoperative pain and neuropathic chronic pain. There is no evidence that magnesium can modulate pain in patients with peripheral arterial occlusive disease (PAOD). We describe the protocol of a single-centre randomised double-blind clinical trial aimed at assessing the efficacy of oral magnesium supplementation in controlling severe pain in patients with advanced PAOD.

Methods and analysis: Adult patients affected by PAOD at stages III and IV of Lèriche-Fontaine classification, who are opioid-naïve, and who have been admitted to our Acute Pain Service for intractable pain, will be eligible. Patients will be randomised to the control group, treated with standard therapy (oxycodone and pregabalin) plus placebo for 2 weeks, or to the experimental group (standard therapy plus magnesium oxide). Patients will be evaluated on days 0, 2, 4, 6, 8, 12 and 14; the following information will being collected: daily oxycodone dose; average and maximum pain (Numerical Rating Scale); pain relief (Pain Relief Scale); characteristics of the pain (Neuropathic Pain Scale); impact of pain on the patient's daily activities (Brief Pain Inventory). The primary outcome will be oxycodone dosage needed to achieve satisfactory analgaesia on day 14. Secondary outcomes will be pain relief on day 2, time needed to achieve satisfactory analgaesia and time needed to achieve a pain reduction of 50%. A sample size calculation was performed for the primary outcome, which estimated a required sample size of 150 patients (75 per group).

Ethics and dissemination: Ethical approval of the study protocol has been obtained from Comitato Etico Provinciale di Brescia, Brescia, Italy. Trial results will be disseminated through scientific journal manuscripts and scientific conference presentations.

Trial registration number: NCT02455726.

Keywords: VASCULAR MEDICINE; VASCULAR SURGERY.

Published by the BMJ Publishing Group Limited. For permission to use (where not already granted under a licence) please go to http://www.bmj.com/company/products-services/rights-and-licensing/

Figures

Figure 1
Figure 1
Study flow will be reported according to the Consolidated Standards of Reporting Trials (CONSORT).

References

    1. Iseri LT, French JH. Magnesium: nature's physiologic calcium blocker. Am Heart J 1984;108:188–93. 10.1016/0002-8703(84)90572-6
    1. Kew JN, Kemp JA. Ionotropic and metabotropic glutamate receptor structure and pharmacology. Psycopharmacology 2005;179:4–29. 10.1007/s00213-005-2200-z
    1. Recio-Pinto E, Castillo C. Peripheral N-methyl-D-aspartate receptors as possible targets for chronic pain treatment. Techn Reg Anesth Pain Manag 2010;14:48–58. 10.1053/j.trap.2010.04.001
    1. Fisher K, Coderre TJ, Hagen NA. Targeting the N-Methyl-D-Aspartate receptor for chronic pain management: preclinical animal studies, recent clinical experience and future research directions. J Pain Symptom Manage 2000;20:358–73. 10.1016/S0885-3924(00)00213-X
    1. Hewitt DJ. The use of NMDA-receptor antagonists in the treatment of chronic pain. Clin J Pain 2000;16:S73–9. 10.1097/00002508-200006001-00013
    1. De Kock MF, Lavand'homme PM. The clinical role of NMDA receptor antagonists for the treatment of postoperative pain. Best Pract Res Clin Anaesthesiol 2007;21:85–98. 10.1016/j.bpa.2006.12.006
    1. Petrenko AB, Yakamura T, Baba H et al. . The role of N-Methyl-D Aspartate NMDA receptors in pain: a review. Anesth Analg 2003;97:1108–16. 10.1213/01.ANE.0000081061.12235.55
    1. Nechifor M. Magnesium involvement in pain. Magnes Res 2011;24:220–2. 10.1684/mrh.2011.0296
    1. Hasanein P, Parviz M, Keshavarz M et al. . Oral magnesium administration prevents thermal hyperalgesia induced by diabetes in rats. Diabetes Res Clin Pract 2006;73:17–22. 10.1016/j.diabres.2005.12.004
    1. Rondón LJ, Privat AM, Daulhac L et al. . Magnesium attenuates chronic hypersensitivity and spinal cord NMDA receptor phosphorylation in a rat model of diabetic neuropathic pain. J Physiol 2010;588(Pt 21):4205–15. 10.1113/jphysiol.2010.197004
    1. Begon S, Pickering G, ESchalier A et al. . Magnesium increases morphine analgesic effect in different experimental models of pain. Anesthesiology 2002;96:627–32. 10.1097/00000542-200203000-00019
    1. Bujalska M, Makulska-Nowak H, Gumulka SW. Magnesium ions and opioid agonists in vincristine-induced neuropathy. Pharmacol Rep 2009;61:1096–104. 10.1016/S1734-1140(09)70172-0
    1. Ulugol A, Aslantas A, Ipci Y et al. . Combined systemic administration of morphine and magnesium sulfate attenuates pain-related behavior in mononeuropathic rats. Brain Res 2002;943:101–4. 10.1016/S0006-8993(02)02618-5
    1. Chen Y, Zhang Y, Zhu YL et al. . Efficacy and safety of an intra-operative intra-articular magnesium/ropivacaine injection for pain control following total knee arthroplasty. J Int Med Res 2009;37:1733–41. 10.1177/147323000903700609
    1. Bondok RS, Abd El-Hady AM. Intra-articular magnesium is effective for postoperative analgesia in arthroscopic knee surgery. Br J Anaesth 2006;97:389–92. 10.1093/bja/ael176
    1. Cizmeci P, Ozkose Z. Magnesium sulphate as an adjuvant to total intravenous anesthesia in septorhinoplasty: a randomized controlled study. Aesth Plast Surg 2007;31:167–73. 10.1007/s00266-006-0194-5
    1. Nechifor M. Magnesium in major depression. Magnes Res 2009;22:S163–6.
    1. Murck H. Ketamine, magnesium and major depression—from pharmacology to pathophysiology and back. J Psychiatr Res 2013;47:955–65. 10.1016/j.jpsychires.2013.02.015
    1. Albrecht E, Kirkham KR, Liu SS et al. . Peri-operative intravenous administration of magnesium sulphate and postoperative pain: a meta-analysis. Anaesthesia 2013;68:79–90. 10.1111/j.1365-2044.2012.07335.x
    1. Seyhan TO, Tugrul M, Sungur MO et al. . Effects of three different dose regimens of magnesium on propofol requirements haemodynamic variables and postoperative pain relief in gynaecological surgery. Br J Anaesth 2006;96:247–52. 10.1093/bja/aei291
    1. Ryu JH, Kang MH, Park KS et al. . Effects of magnesium sulphate on intraoperative anaesthetic requirements and postoperative analgesia in gynaecology patients receiving total intravenous anaesthesia. Br J Anaesth 2008;100:397–403. 10.1093/bja/aem407
    1. Tramer MR, Schneider J, Marti RA et al. . Role of magnesium sulfate in postoperative analgesia. Anesthesiology 1996;84:340–7. 10.1097/00000542-199602000-00011
    1. Song JW, Lee YW, Yoon KB et al. . Magnesium sulfate prevents remifentanil-induced postoperative hyperalgesia in patients undergoing thyroidectomy. Anesth Analg 2011;113:390–7. 10.1213/ANE.0b013e31821d72bc
    1. Tugrul S, Degirmenci N, Eren SB et al. . Analgesic effect of magnesium in post-tonsillectomy patients: a prospective randomised clinical trial. Eur Arch Otorhinolatyngol 2015;272:2483–7. 10.1007/s00405-014-3219-8
    1. Borazan H, Kececioglu A, Okesli S et al. . Oral magnesium lozenge reduces postoperative sore throat: a randomized, prospective, placebo-controlled study. Anesthesiology 2012;117:512–18. 10.1097/ALN.0b013e3182639d5f
    1. Brill S, Sedgwick PM, Hamann W et al. . Efficacy of intravenous magnesium in neuropathic pain. Br J Anaesth 2002;89:711–14. 10.1093/bja/89.5.711
    1. Crosby V, Wilcock A, Corcoran R. The safety and efficacy of a single dose (500 mg or 1 g) of intravenous magnesium sulfate in neuropathic pain poorly responsive to strong opioid analgesics in patients with cancer. J Pain Symptom Manage 2000;19:35–9. 10.1016/S0885-3924(99)00135-9
    1. Suresh S, Lozono S, Hall SC. Large-dose intravenous methotrexate-induced cutaneous toxicity: can oral magnesium oxide reduce pain? Anesth Analg 2003;96:1413–14. 10.1213/01.ANE.0000057140.33744.65
    1. Kroin JS, Mc Carthy RJ, Von Roenn N et al. . Magnesium sulfate potentiates morphine antinociceptoin at the spinal level. Anesth Analg 2000;90:913–17. 10.1213/00000539-200004000-00025
    1. Yousef AA, Al-deeb AE. A double-blinded randomized controlled study of the value of sequential intravenous and oral magnesium therapy in patients with chronic low back pain with a neuropathic component. Anaesthesia 2013;68:260–6. 10.1111/anae.12107
    1. Lysakowski C, Dumont L, Czarnetzki C et al. . Magnesium as an adiuvant to postoperative analgesia: a systematic review of randomized trials. Anesth Analg 2007;104:1532–9. 10.1213/
    1. Pickering G, Morel V, Simen E et al. . Oral magnesium treatment in patients with neuropathic pain: a randomized clinical trial. Magnes Res 2011;24:28–35. 10.1684/mrh.2011.0282
    1. Felsby S, Nielsen J, Arendt-Nielsen L et al. . NMDA receptor blockade in chronic neurophatic pain: a comparison of ketamine and magnesium chloride. Pain 1996;64:283–91. 10.1016/0304-3959(95)00113-1
    1. Mikkelsen S, Dirks J, Fabricius P et al. . Effect of intravenous magnesium on pain and secondary hyperalgesia associated with the heat/capsaicin sensitization model in healthy volunteers. Br J Anaesth 2001;86:871–3. 10.1093/bja/86.6.871
    1. Rüger LJ, Irnich D, Abahji TN et al. . Characteristics of chronic ischemic pain in patients with peripheral arterial disease. Pain 2008;139:201–8. 10.1016/j.pain.2008.03.027
    1. Shechter M, Merz NB, Stuehlinger HG et al. . Effects of oral magnesium therapy on exercise tolerance, exercise-induced chest pain and quality of life in patients with coronary artery disease. Am J Cardiol 2003;91:517–21. 10.1016/S0002-9149(02)03297-6
    1. Fontaine R, Kieny R, Gangloff JM et al. . Long-term results of restorative arterial surgery in obstructive diseases of the arteries. J Cardiovasc Surg 1964;5:463–72.
    1. Caraceni A, Cherny N, Fainsinger R et al. . Pain measurement tools and methods in clinical research in palliative care: recommendations of an expert working group of the European Association of Palliative Care. J Pain and Symptom Manage 2002;23:239–55. 10.1016/S0885-3924(01)00409-2
    1. Frampton CL, Hughes-Webb P. The measurement of pain. Clin Oncol 2011;23:381–6. 10.1016/j.clon.2011.04.008
    1. Galer BS, Jensen MP. Development and preliminary validation of a pain measure specific to neuropathic pain: the Neuropathic Pain Scale. Neurology 1997;48:332–8. 10.1212/WNL.48.2.332
    1. Bonezzi C, Bettaglio R, Jurusich D et al. . Scala del Dolore Neuropatico, versione italiana di The Neuropathic Pain Scale. Minerva Anestesiol 2000;66:841–3.
    1. Negri E, Bettaglio R, Demartini L et al. . Validation if Italian version of the Neuropathic Pain Scale and its clinical implications. Minerva Anestesiol 2002;68:95–104.
    1. Charlson ME, Pompei P, Ales KL et al. . A new method of classifying prognostic comorbidity in longitudinal study: development and validation. J Chron Dis 1987;40:373–83. 10.1016/0021-9681(87)90171-8
    1. Charlson ME, Szatrowski TP, Peterson J et al. . Validation of a combined comorbidity index. J Clin Epidemiol 1994;47:1245–51. 10.1016/0895-4356(94)90129-5
    1. Benyamin R, Trescot AM, Datta S et al. . Opioid complications and side effects. Pain Physician 2008;11:S105–20.

Source: PubMed

3
Abonnieren